US10583171B2 - NMDAR antagonists for the treatment of diseases associated with angiogenesis - Google Patents

NMDAR antagonists for the treatment of diseases associated with angiogenesis Download PDF

Info

Publication number
US10583171B2
US10583171B2 US15/779,369 US201615779369A US10583171B2 US 10583171 B2 US10583171 B2 US 10583171B2 US 201615779369 A US201615779369 A US 201615779369A US 10583171 B2 US10583171 B2 US 10583171B2
Authority
US
United States
Prior art keywords
nmdar
angiogenesis
antagonist
mice
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US15/779,369
Other languages
English (en)
Other versions
US20180318389A1 (en
Inventor
Sylvia Cohen-Kaminsky
Sebastien Dumas
Gilles Bru-Mercier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institut National de la Sante et de la Recherche Medicale INSERM
Universite Paris Sud Paris 11
Original Assignee
Institut National de la Sante et de la Recherche Medicale INSERM
Universite Paris Sud Paris 11
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut National de la Sante et de la Recherche Medicale INSERM, Universite Paris Sud Paris 11 filed Critical Institut National de la Sante et de la Recherche Medicale INSERM
Assigned to UNIVERSITE PARIS-SUD, INSERM (INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE) reassignment UNIVERSITE PARIS-SUD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COHEN-KAMINSKY, SYLVIA, BRU-MERCIER, Gilles, DUMAS, SEBASTIEN
Publication of US20180318389A1 publication Critical patent/US20180318389A1/en
Application granted granted Critical
Publication of US10583171B2 publication Critical patent/US10583171B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1787Receptors; Cell surface antigens; Cell surface determinants for neuromediators, e.g. serotonin receptor, dopamine receptor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/451Non condensed piperidines, e.g. piperocaine having a carbocyclic group directly attached to the heterocyclic ring, e.g. glutethimide, meperidine, loperamide, phencyclidine, piminodine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to a method of treating disease associated with angiogenesis using an N-Methyl-D-aspartate receptor (NMDAR) antagonist. More specifically, it concerns use of an NMDA receptor antagonist, for the treatment of diseases associated with angiogenesis such as tumor angiogenesis, ocular neovascular disease, age-related macular degeneration (AMD).
  • N-Methyl-D-aspartate receptor AMD
  • Angiogenesis is a biological process of generating new blood vessels into a tissue or an organ. Under normal physiological conditions, humans or animals undergo angiogenesis only in very specific restricted situations. For example, angiogenesis is normally observed in wound healing, fetal and embryonic development and formation of the corpus luteum, endometrium and placenta. It is widely accepted that new vessel growth is tightly controlled by many angiogenic regulators and the switch of the angiogenesis phenotype depends on the net balance between up-regulation of angiogenic stimulators and down-regulation of angiogenic suppressors. The control of angiogenesis has been found to be altered in certain disease states and, in many cases, the pathological damage associated with the disease is related to the uncontrolled angiogenesis.
  • Angiogenesis begins with the erosion of the basement membrane by enzymes released by endothelial cells and leukocytes. The endothelial cells, which line the lumen of blood vessels, then protrude through the basement membrane. Angiogenic stimulants induce the endothelial cells to migrate through the eroded basement membrane. The migrating cells form a “sprout” off the parent blood vessel, where the endothelial cells undergo mitosis and proliferate. The endothelial sprouts merge with each other to form capillary loops, creating the new blood vessel.
  • angiostatin which consists of the plasminogen kringle domains I through IV.
  • apolipoprotein (a) one of the proteins having kringle structures, is a candidate for a novel angiogenesis inhibitor.
  • Taylor et al. have used protamine to inhibit angiogenesis, although its toxicity limits its practical use as a therapeutic agent (Taylor et al., 1982, Nature, Vol. 297 : 307).
  • Folkman et al. have disclosed the use of heparin and steroids to control angiogenesis (Folkman et al., 1983, Science, Vol. 221 : 719).
  • Other agents which have been used to inhibit angiogenesis include ascorbic acid ethers and related compounds (Japanese Patent Kokai Tokkyo Koho No 58-131978).
  • a fungal product, fumagillin is a potent angiostatic agent in vitro, as well as its synthetic derivative O-substituted fumagillin.
  • Bevacizumab is a humanized antibody directed against the angiogenic factor VEGF. Bevacizumab prevents the binding of VEGF to its effector receptor and has been initially used for treating colorectal cancer.
  • N-methyl-D-Aspartate Receptor is a ligand-gated and voltage-dependent channel belonging to the ionotropic glutamate receptor family 5 . It forms an heterotetrameric ion channel across the cell membrane and is composed of two GluN1 subunits (encoded by the GRIN1 gene), obligatory subunits to form functional NMDAR, containing a glycine or D-serine binding site, mainly associated to two GluN2 subunits either GluN2A, GluN2B, GluN2C or GluN2D subunits comprising the glutamate binding site, and modulating channel properties 5 .
  • NMDAR central nervous system
  • CNS central nervous system
  • NMDAR distributes at the cell membrane in cell-cell contact sites either synaptic (defined by a close contact between neurons processes) or extrasynaptic sites (associated to close contacts mainly between neurons and microglial cells or astrocytes) 6 .
  • Dysregulation of the NMDAR-mediated glutamatergic communication has been pointed out in many disorders in the CNS i.e.
  • NMDAR neurodegenerative disorders Alzheimer, Parkinson and Huntington diseases 7 , but also depression, anxiety 8 , schizophrenia, autism 9 , stroke 10 etc. characterized by transcriptional and/or post-translational modification of NMDAR 10,11 .
  • NMDAR has also been found outside the CNS: it is expressed by osteoclasts, osteoblasts, pancreatic beta cells, testis, keratinocytes, renal podocytes, immune cells, skeletal muscle, cardiomyocyte, etc. and also cerebral and aortic endothelial and smooth muscle cells 12-25 .
  • Vgluts vesicular glutamate transporters
  • peripheral tissues such as bones, islets of Langerhans, testes, pineal gland, intestines and stomach 26 .
  • Studies have suggested a potential role of NMDAR in the development of chronic peripheral disorders such as osteoporosis 13 and type 2 diabetes mellitus 15 , indicating that targeting peripheral NMDAR with a specific antagonist could be beneficial in these conditions.
  • Some cancer cells can hijack the NMDAR to proliferate in an aberrant way 27 .
  • NMDAR-specific antagonists in animal models of cancer has shown dramatic improvements of the animal survival breaking tumor growth 28,29 . Additionally it has been demonstrated that NMDAR activation of cerebral or aortic endothelial cells contributes to blood-barrier opening, monocyte infiltration, reactive oxygen species production, apoptosis or proliferation 18-24 . In aortic smooth muscle cells, NMDAR activation triggers proliferation and MMP-2 synthesis 25 .
  • the invention provides an antagonist of the N-Methyl-D-aspartate receptor (NMDAR), for inhibiting angiogenesis.
  • NMDAR N-Methyl-D-aspartate receptor
  • the antagonist of NMDAR according to the invention is for use in treating angiogenic dependent or angiogenic associated disease.
  • said NMDAR antagonist may be a low molecular weight compound, e. g. a small organic molecule (natural or not).
  • said antagonist is a competitive or a non-competitive or an uncompetitive antagonist of the NMDA receptor.
  • the angiogenic dependent or angiogenic associated disease according to the invention is selected from tumor angiogenesis, ocular neovascular disease.
  • the antagonist according to the invention includes:
  • an anti-NMDAR antibody or antibody fragment that may partially or completely block NMDAR activation by glutamate (and glycine);
  • a polypeptide that may partially or completely inhibit trafficking and anchoring at the cell membrane of the NMDAR.
  • the invention provides an inhibitor of NMDAR expression for inhibiting angiogenesis.
  • PAH pulmonary arterial hypertension
  • the inventors provide evidence that NMDAR activation promotes pulmonary arterial remodeling in PAH.
  • Using different approaches including in situ observations of dysregulated features of glutamatergic communication in human explanted PAH lungs, in vitro evidence of glutamate release from vascular cells and the role of NMDAR activation in vascular cell proliferation and angiogenesis, in vivo targeted knockout of NMDAR in endothelial or smooth muscle cells, they demonstrate a role for NMDAR activation in the vascular remodeling processes underlying pulmonary hypertension.
  • the inventors show that blocking the NMDA receptor constitutes an alternative therapeutic axis in a disease associated with vascular cell proliferation and misguided and uncontrolled angiogenesis.
  • the present invention provides methods and compositions (such as pharmaceutical compositions) for treating diseases associated with angiogenesis.
  • an object of the invention is an antagonist of the N-Methyl-D-aspartate receptor (NMDAR), for inhibiting angiogenesis.
  • N-Methyl-D-aspartate receptor N-Methyl-D-aspartate receptor
  • the antagonist of NMDAR according to the invention is for use in treating angiogenic dependent or angiogenic associated disease.
  • the NMDAR antagonist for use in treating disease associated with angiogenesis.
  • the invention relates to a method of inhibiting angiogenesis in a subject in need thereof comprising administering an antagonist of NMDAR.
  • the invention relates to a method of treating angiogenic dependent or angiogenic associated disease in a subject in need thereof comprising administering an antagonist of NMDAR.
  • antagonist a natural or synthetic compound that has a biological effect opposite to that of an agonist.
  • An antagonist binds the receptor and blocks the action of a receptor agonist by competing with the agonist for receptor.
  • An antagonist is defined by its ability to block the actions of an agonist.
  • compound as used throughout the specification includes but is not limited to: small organic molecule; antibody or antibody fragment that may partially or completely block NMDAR activation by glutamate (and glycine), a polypeptide or an inhibitor of NMDAR expression.
  • NMDAR N-Methyl-D-aspartate receptor
  • NMDA receptors consist of two glycine-binding GluN1 subunits encoded by eight splice variants of a single gene and two glutamate-binding GluN2 (A-D) subunits, arising from four genes (Traynelis S F et al Pharm Rev. 2010; 62(3)).
  • NMDAR may also have a GluN3 (A-B) subunit Kehoe L A et al Neural Plast.
  • NMDA receptors uniquely require both glutamate and glycine as co-agonists for receptor activation (Johnson J W, et al Nature. 1987). Following channel opening, membrane depolarization is required to relieve the voltage-dependent Mg2+ block before ions can permeate the channel pore (Mayer M L, Nature. 1984).
  • NMDA receptor antagonist refers to a compound that reduces or block, the flow of cations (Na+, K+, Ca2+) through the NMDA receptor.
  • the NMDA receptor antagonists comprise four categories of compounds: competitive antagonists, which bind to and block the binding site of the neurotransmitter glutamate; glycine antagonists, which bind to and block the glycine site; non-competitive antagonists, which inhibit NMDA receptors by binding to allosteric sites; and uncompetitive antagonists or channel blockers, which block the ion channel by binding to a site within it.
  • the antagonist of the NMDA receptor binds to the NMDA receptor and blocks the biological effect of glutamate (and glycine) on NMDAR.
  • an antagonist can act by occupying the ligand binding site or a portion thereof of the NMDA receptor, thereby making the receptor inaccessible to its natural ligand so that its normal biological activity is prevented or reduced.
  • a test based on the effect of the NMDAR antagonist candidate on the inhibition of angiogenesis (Matrigel assay) as explained in the examples ( FIG. 1 ) may be used.
  • a NMDAR antagonist can act by binding directly to the intracellular domain of the receptor and inhibiting NMDAR trafficking to the cell membrane.
  • a NMDAR antagonist may for instance block or inhibit NMDAR activation and/or trafficking to the cell membrane and/or anchoring at the cell membrane: e.g., blocking or inhibiting interaction of intracellular domain of the NMDAR with PDZ domain of scaffolding protein (like PSD95) and prevent its anchoring to the cell membrane (Tymianski M. et al Science. 2002 Oct. 25; 298(5594):846-50).
  • the NMDAR antagonist is a low molecular weight compound, e. g. a small organic molecule (natural or not).
  • small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals.
  • Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
  • the NMDA receptor antagonist according to the invention is a small organic molecule that binds any part of the NMDAR, including the channel part, and be able through allosteric effects, to modulate NMDAR activity.
  • Low molecular weight NMDAR antagonists are well known in the art.
  • low molecular weight NMDAR antagonists that may be used by the invention include, for example competitive NMDAR antagonists; glycine NMDAR antagonists; non-competitive NMDAR antagonists, and uncompetitive NMDAR antagonists or channel blockers as well as all pharmaceutically acceptable salts and solvates of said NMDAR antagonists, such as those described in the following patent publications: International and national Patent Publication Nos.
  • low molecular weight NMDAR antagonists include any of the channel blockers (such as memantine; lanicemine, Remacemide amantadine; tiletamine; rimantadine, phencyclidine (PCP); PCP hydrochloride functional derivatives; dizocilpine MK-801, Argiotoxin 636, dextrorphan) since these channel blockers have also been described in Yamakura T, Neuroreport 1993; 4(6):687-90, Kashiwagi K, Mol Pharm 2002; 61(3):533-45 LePage K T Neuropharmacology 2005; 49(1):1-16, Jin L, J Pharmacol Exp Ther 2007; 320(1):47-55, Burnashev N, Science 1992; 257(5075):1415-19.
  • channel blockers such as memantine; lanicemine, Remacemide amantadine; tiletamine; rimantadine, phencyclidine (PCP); PCP hydrochloride functional derivatives; d
  • NMDAR antagonist that can be used according to the present invention may be the (3,5-dimethyladamantan-1-amine) (also known as Memantine, Axura, Namenda, Ebixa and Mimetix) (Patent Publication No. U.S. Pat. No. 5,061,703, EP0392059) and which have the following structure:
  • NMDAR antagonist may be the [5R,10S]-[+]-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine (also known as MK801, Dizocilpine (INN)) (Patent Publication No. U.S. Pat. No. 4,399,141, GB2061947) and which have the following structure:
  • a low molecular weight NMDAR antagonist that can be used according to the present invention may be the adamantan-1-amine (also known as Amantadine, Symmetrel) (Patent Publication No. U.S. Pat. No. 3,310,469, WO1992000066)
  • NMDAR antagonist may be the 1-(1-phenylcyclohexyl)piperidine (also known as Phencyclidine, PCP, Sernyl)) (Patent Publication No. UK836083 and U.S. Pat. No. 3,097,136) and which have the following structure:
  • NMDAR antagonist may be the (RS)-2-(2-Chlorophenyl)-2-(methylamino)cyclohexanone (also known as Ketamine and Ketamine hydrochloride) (Patent Publication No. U.S. Pat. No. 3,254,124) and which have the following structure:
  • a low molecular weight NMDAR antagonist that can be used according to the present invention may be the 1(1-(1-aminoethyl)adamantane) (also known as rimantadine, Flumadine)) (Patent Publication No. U.S. Pat. Nos. 4,551,552 and 3,352,912) and which have the following structure:
  • NMDAR antagonists may be the 4-[2-(4-benzylpiperidin-1-yl)-1-hydroxypropyl]phenol (also known as ifenprodil, Vadilex; Dilvax; Creocral;)) (Patent Publication No. U.S. Pat. No. 3,509,164 and EP0109317) and which have the following structure:
  • NMDAR antagonists that can be used according to the present invention may be the 2-amino-5-phosphonopentano ⁇ c acid (UPAC nomenclature) (also known as APV or AP5 or 2-amino-5-phosphonovaleric acid) which inhibits the binding site and which has the following structure
  • UPAC nomenclature also known as APV or AP5 or 2-amino-5-phosphonovaleric acid
  • NMDA receptor antagonists include but are not limited to any of the NMDA receptor antagonists described in Santagelo R M. et al. (Exp Opin. Ther Pat 22(11) 2012) or described in Strong KL and al ((Exp Opin. Ther Pat 24(12) 2014) or Mlodzic H et al (Exp Opin. Ther Pat 6(4), 1996) or Kolher M. et al (Exp Opin. Ther Pat 20(12), 2010) or Kulagowski JJ (Exp Opin. Ther Pat 6(10), 1996) which are herein incorporated by reference.
  • the low molecular weight antagonist is non-competitive antagonist of the NMDA receptor.
  • non-competitive antagonists include but are not limited to: ketamine, HU-211 or Dexanabinol, Rhynchophylline aptiganel or Cerestat or CNS-5 1102, ifenprodil, and/or analogs and/or functional derivatives thereof.
  • the low molecular weight antagonist is an uncompetitive channel blocker of the NMDA receptor.
  • uncompetitive antagonists of the NMDA receptor include but are not limited to: memantine; lanicemine, Remacemide amantadine; tiletamine; rimantadine, phencyclidine (PCP); PCP hydrochloride functional derivatives; dizocilpine MK-801, Argiotoxin 636, dextrorphan and/or analogs and/or functional derivatives thereof.
  • said compound is memantine, or MK-801 and/or an analog and/or a functional derivative thereof.
  • the low molecular weight antagonist is a competitive channel blocker of the NMDA receptor.
  • competitive antagonists of the NMDA receptor include but are not limited to: AP5 (also known as APV), AP7, CPPene and Selfotel also known as CGS 19755), and/or analogs and/or functional derivatives thereof.
  • Analog refers broadly to the modification or substitution of one or more chemical moieties on a parent compound and may include functional derivatives, positional isomers, tautomers, zwitterions, enantiomers, diastereomers, racemates, isosteres or stereochemical mixtures thereof.
  • the term “Functional derivative” refers to a compound which possesses similar IC50 values and kinetics properties as MK-801 and memantine to NMDA receptor.
  • the functional derivative of the invention possesses the capacity to suppress and/or decrease the angiogenesis.
  • the functional derivative of the invention also may not target the central NMDAR, by not crossing the blood brain barrier after administration of the compound.
  • the antagonist of the invention does not cross the blood brain barrier.
  • antagonists include but are not limited to AP5, also known as APV ((2R)-amino-5-phosphonovaleric acid; (2R)-amino-5-phosphonopentanoate) (Synaptic plasticity and learning: selective impairment of learning rats and blockade of long-term potentiation in vivo by the N-methyl-D-aspartate receptor antagonist AP5. Morris R G Journal of Neuroscience. 1989 September; 9(9):3040-57. PMID 2552039) and L-703,717 (Allosteric modulation of the glutamate site on the NMDA receptor by four novel glycine site antagonists.
  • the NMDAR antagonist consists in an antibody or antibody fragment that may partially or completely block NMDA receptor activation by glutamate and glycine, or modulate its activity by binding to the channel site or any allosteric site of the receptor.
  • Antibodies against NMDAR are known in the art.
  • Example of antibody-based NMDAR antagonists include those described in WO 2014187879 and include the well characterized antibody NMDA NR 1 Pan Antibody, mouse monoclonal, Novus biologicals, NB 300-118.
  • Additional antibody antagonists may be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • Various adjuvants known in the art may be used to enhance antibody production.
  • antibodies useful in practicing the invention may be polyclonal, monoclonal antibodies are preferred.
  • Monoclonal antibodies against NMDA receptor may be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture.
  • Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975); the human B-cell hybridoma technique (Cote et al., 1983); and the EBV-hybridoma technique (Cole et al, 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • techniques described for the production of single chain antibodies may be adapted to produce anti-NMDAR, or anti-NMDAR single chain antibodies.
  • NMDAR antagonists useful in practicing the present invention also include anti-NMDAR, or anti-NMDAR antibody fragments including but not limited to F(ab′).sub.2 fragments, which may be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which may be generated by reducing the disulfide bridges of the F(ab′).sub.2 fragments.
  • F(ab′).sub.2 fragments which may be generated by pepsin digestion of an intact antibody molecule
  • Fab fragments which may be generated by reducing the disulfide bridges of the F(ab′).sub.2 fragments.
  • Fab and/or scFv expression libraries may be constructed to allow rapid identification of fragments having the desired specificity to NMDA receptor.
  • Humanized anti-NMDAR and antibody fragments therefrom may also be prepared according to known techniques.
  • “Humanized antibodies” are forms of non-human (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the NMDAR antagonist consists in a polypeptide.
  • a polypeptide is a chain of amino acids linked by peptide bonds which contains between 10 and 100 amino acids.
  • the antagonist of the invention may block NMDA receptor-protein interaction such as PSD-95 (Postsynaptic density-95 protein) which interacts with an intracellular domain of the NMDAR and prevents its anchoring to the cell membrane (Tymianski M. et al Science. 2002 Oct. 25; 298(5594):846-50).
  • PSD-95 Postsynaptic density-95 protein
  • polypeptide targeting intracellular domain of the NMDAR that can be used according to the present invention include those described in the US20110097324; U.S. Pat. Nos. 8,071,548; 8,008,253; 7,846,897; US20120083449 US20100160240, U.S. Pat. No. 7,510,824.
  • Another object of the invention is an inhibitor of NMDAR expression for inhibiting angiogenesis.
  • Inhibitors of NMDAR expression for use in the present invention may be based on antisense oligonucleotide constructs.
  • Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, act to directly block the translation of NMDAR mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of NMDAR proteins, and thus activity, in a cell.
  • siRNAs targeting NMDAR that can be used according to the present invention include those described in the US Patent Publication No. U.S. Pat. No. 8,372,817.
  • Small inhibitory RNAs may also function as inhibitors of NMDAR expression for use in the present invention.
  • NMDAR gene expression may be reduced by contacting the pathological site with deregulated angiogenesis like tumor, cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that NMDAR expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschi, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, G J.
  • Ribozymes may also function as inhibitors of NMDAR expression for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of NMDAR mRNA sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GuU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features, such as secondary structure, that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • antisense oligonucleotides and ribozymes useful as inhibitors of NMDAR expression may be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules may be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention may be introduced as a means of increasing intracellular stability and half-life.
  • Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5′ and/or 3′ ends of the molecule, or the use of phosphorothioate or 2′-O-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
  • Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a “vector” is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing NMDAR.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • adenovirus adeno-associated virus
  • SV40-type viruses polyoma viruses
  • Epstein-Barr viruses Epstein-Barr viruses
  • papilloma viruses herpes virus
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • Standard protocols for producing replication-deficient retroviruses including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles
  • KRIEGLER A Laboratory Manual,” W.H. Freeman C.O., New York, 1990
  • MURRY Methodhods in Molecular Biology,” vol. 7, Humana Press, Inc., Cliffton, N.J., 1991.
  • adeno-viruses and adeno-associated viruses are double-stranded DNA viruses that have already been approved for human use in gene therapy.
  • the adeno-associated virus may be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
  • the adeno-associated virus may integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno-associated virus may also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g., SANBROOK et al., “Molecular Cloning: A Laboratory Manual,” Second Edition, Cold Spring Harbor Laboratory Press, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, may express a peptide from a gene operatively encoded within the plasmid.
  • Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid may be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • Another object of the invention relates to a method for inhibiting angiogenesis or method for treating disease associated with angiogenesis comprising administering a subject in need thereof with a therapeutically effective amount of an antagonist or inhibitor of expression as described above.
  • treating means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • the term “patient” or “patient in need thereof”, is intended for a human or non-human mammal affected or likely to be affected with a pathological condition or disease involving deregulated angiogenesis.
  • a “therapeutically effective amount” of the antagonist or inhibitor of expression as above described is meant a sufficient amount of the antagonist or inhibitor of expression to treat disease associated with angiogenesis at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidential with the specific therapeutical agent employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • Antagonists of the invention may further be identified by the screening methods described in the state of the art.
  • the screening methods of the invention may be carried out according to known methods.
  • the screening method may measure the binding of a candidate compound to the NMDA receptor, or to cells or membranes bearing the NMDAR receptor, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound.
  • a screening method may involve measuring or, qualitatively or quantitatively, detecting the competition of binding of a candidate compound to the receptor with a labelled competitor (e.g., antagonist or agonist). Further, screening methods may test whether the candidate compound results in a signal generated by an antagonist of the receptor, using detection systems appropriate to cells bearing the NMDA receptor.
  • Antagonists may be assayed in the presence of a known agonist (e.g., glutamate or NMDA) and co-agoniste (glycine, D-serine) and an effect on activation by the agonist by the presence of the candidate compound is observed.
  • a known agonist e.g., glutamate or NMDA
  • co-agoniste glycine, D-serine
  • competitive binding using known agonist such glutamate (or glycine) is also suitable.
  • an object of the invention is an antagonist of the N-Methyl-D-aspartate receptor (NMDAR), for inhibiting angiogenesis.
  • N-Methyl-D-aspartate receptor N-Methyl-D-aspartate receptor
  • the antagonist of NMDAR according to the invention is for use in treating angiogenic dependent or angiogenic associated disease.
  • disease associated with angiogenesis means a disease associated with a persistent, unregulated angiogenesis.
  • the conditions or diseases that are concerned are those for which angiogenesis is pathological and should be reduced or blocked.
  • Persistent, unregulated angiogenesis occurs in a multiplicity of disease states, including tumor growth and tumor metastasis, and supports the pathological damage seen in these conditions.
  • the diverse pathological states that are due to unregulated angiogenesis have been grouped together as angiogenic dependent or angiogenic associated diseases or conditions.
  • ocular neovascular disease is characterized by invasion of new blood vessels into the structure of the eye such as the retina or the cornea. It is the most common cause of blindness and is involved in approximately twenty eye diseases.
  • age-related macular degeneration the associated visual problems are caused by an ingrowth of chorioidal capillaries through defects in Bruch's membrane with proliferation of fibrovascular tissues beneath the retinal pigment epithelium.
  • Angiogenic damage is also associated with diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, neovascular glaucoma, and retrolental fibroplasias.
  • corneal neovascularization include, but are not limited to, epidemic keratocunjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium kreatitis sicca, sjogrens, acne rosacea, phylectenulosis, syphilis, mycobacterial infections, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infections, Herpes zoster infections, protozoan infections, Kaposi's sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, rheumatois arthritis, systemic lupus, polyarteritis, Wegener's sarcoidiosis, scleritis, Stevens-Johnson disease, pemphigoid, radial keratotomy and corneal graft rejection.
  • Diseases associated with retinal/choroidal neovascularization include, but are not limited to, diabetic retinopathy, macular degeneration, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum, Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, mycobaterial infections, Lyme's disease, systemic lupus erythematosis, retinopathy of prematurity, Eale's disease, Bechet's disease, infections causing retinitis or choroiditis, presumed ocular histoplasmosis, Best's disease, myopia, optic pits, Stargardt's disease, pars planitis, chronic etinal detachment, hyperviscosity syndromes, toxoplasmosis, trauma and post-laser complications.
  • Other diseases include, but are not limited to, diseases
  • angiogenesis Another disease in which angiogenesis is involved is rheumatoid arthritis.
  • the endothelial cells release factors and reactive oxygen species that lead to panus growth and cartilage destruction.
  • the factors involved in angiogenesis may actively contribute to, and help maintain, the chronically inflamed state of rheumatoid arthritis.
  • Angiogenesis is also involved in osteoarthritis.
  • the activation of the chondrocytes by angiogenic-related factors contributes to the destruction of the joint.
  • deregulation of angiogenesis is the cause of hemangioma, which is one of the most frequent angiogenic diseases in childhood.
  • Deregulation of angiogenesis is also responsible for damage found in hereditary diseases such as Osler-Weber-Rendu disease, or hereditary hemorrhagic telangiectasia. This is an inherited disease characterized by multiple small angiomas, tumors of blood or lymph vessels.
  • Angiogenesis is also involved in normal physiological processes such as reproduction and wound healing. Angiogenesis is an important step in ovulation and also in implantation of the blastula after fertilization. Prevention of angiogenesis could be used to induce amenorrhea, to block ovulation or to prevent implantation by the blastula.
  • both primary and metastatic tumors need to recruit angiogenic vessels for their growth. If this angiogenic activity could be repressed or eliminated, then the tumor would not grow. Thus, angiogenesis is prominent in solid tumor formation and metastasis.
  • Angiogenic factors have been found associated with various solid tumors such as rhabdomyosarcomas, retinoblastoma, Ewing sarcoma, neuroblastoma, osteosarcoma as well as with colorectal cancer.
  • Tumors in which angiogenesis is important include solid tumors, and benign tumors such as acoustic neurona, neurofibroma, trachoma and pyogenic granulomas.
  • angiogenesis has been associated with blood-born tumors such as leukemias, any of various acute or chronic neoplastic diseases of the bone marrow in which unrestrained proliferation of white blood cells.
  • Angiogenesis is important in two stages of tumor metastasis.
  • the first stage where angiogenesis is important is in the vascularization of the tumor which allows tumor cells to enter the blood stream and to circulate throughout the body. After the tumor cells have left the primary site, and have settled into the secondary, metastasis site, angiogenesis must occur before the new tumor can grow and expand.
  • deregulation of angiogenesis is the cause of a wide variety of pathological conditions or disease states.
  • the antagonist or inhibitor of expression of the invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions for use in the treatment of pathological conditions with deregulated angiogenesis as illustrated here above.
  • “Pharmaceutically” or “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • the active principle in the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, may be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the antagonist or inhibitor of expression of the invention may be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the carrier may also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms may be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions may be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • solutions are administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like may also be employed.
  • aqueous solutions for parenteral administration in an aqueous solution
  • the solution is suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which may be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • the antagonist or inhibitor of expression of the invention may be formulated within a therapeutic mixture to comprise about 0.0001 to 1.0 milligrams, or about 0.001 to 0.1 milligrams, or about 0.1 to 1.0 or even about 10 milligrams per dose or so. Multiple doses may also be administered.
  • parenteral administration such as intravenous or intramuscular injection
  • other pharmaceutically acceptable forms include, e.g. tablets or other solids for oral administration; liposomal formulations; time release capsules; and any other form currently used.
  • FIG. 1 Activation of endothelial cell NMDAR contributes to vascular remodeling in the development of pulmonary hypertension.
  • FIG. 2 Supplementary figures about activation of endothelial cell NMDAR contributes to vascular remodeling in the development of pulmonary hypertension.
  • hPASMC human pulmonary arterial smooth muscle cells
  • hPMVEC human pulmonary microvascular endothelial cells
  • P4 and P7 both from healthy non-smoker and non-alcoholic donors and on primary culture of hippocampal neurons from rat fetuses.
  • hPASMC culture 250 000 cells were seeded in T75 flasks (BD Falcon, CORNING, Tewksbury, Mass., USA) and grown in the SmGm2 complete medium containing SmBm basal medium and also basic fibroblast growth factor, epidermal growth factor, insulin and gentamycin/amphotericin 1 ⁇ (all from LONZA, Basel, Switzerland).
  • hPMVEC culture 375 000 cells were seeded in T75 flasks (BD Falcon, CORNING, Tewksbury, Mass., USA) and grown in the EGM2-MV complete medium containing EBM2 basal medium and basic fibroblast growth factor-b, epidermal growth factor, vascular endothelial growth factor, hydrocortisone, ascorbic acid, insulin-like growth factor and gentamycin/amphotericin 1 ⁇ (all from LONZA, Basel, Switzerland).
  • EGM2-MV complete medium containing EBM2 basal medium and basic fibroblast growth factor-b, epidermal growth factor, vascular endothelial growth factor, hydrocortisone, ascorbic acid, insulin-like growth factor and gentamycin/amphotericin 1 ⁇ (all from LONZA, Basel, Switzerland).
  • All cells were cultured at 37° C. in a humidified atmosphere of 5% CO 2 and 95% air.
  • Matrigel assays were performed on ⁇ -slide angiogenesis (IBIDI, Martinsried, Germany). 100 ⁇ L of phenol-red free matrigel matrix (BD, Franklin lakes, N.J., USA) were put in each wells and slides were incubated during 30 min at 37° C. Then, 50 ⁇ L of EGM2-MV medium with our without NMDAR antagonists (100 ⁇ M (+)-MK-801 maleate or 50 ⁇ M DAP-V, both from ABCAM BIOCHEMICALS, Cambridge, UK) were added on matrigel gels during 1 hour. 3500 hPMVEC were seeded in each well in presence or absence of NMDAR antagonists.
  • IBIDI phenol-red free matrigel matrix
  • hPASMC/hPMVEC co-culture model of angiogenesis 20000 PASMC were seeded on glass coverslips in a 12-well plate (CORNING, Tewksbury, Mass., USA) and cultured until confluence in SmGm2 complete medium. After reaching high confluence, 80000 PMVEC were seeded on the top of the PASMC layer switching the medium from SmGm2 complete medium to a modified EGM2-MV complete medium containing 2% FBS with our without NMDAR antagonist MK-801 at 0 ⁇ M, 10 ⁇ M, 30 ⁇ M or 100 ⁇ M MK-801 (6 replicates per condition).
  • mice strain used are B6.129S4-Grin1 tm2Stl /J (further named as GRIN1 fl/fl mice), (from JACKSON LABORATORY, Bar Harbor, Me., USA) and B6.Cg-Tg(Tek-cre/ERT2)1Arnd/ArndCnrm (further named as Tek-cre mice) (EUROPEAN MOUSE MUTANT ARCHIVE, CNR Monterotondo, Monterotondo, RM, Italy). Briefly, GRIN1 fl/fl mice were crossed with Tek-cre mice.
  • mice were used as controls after 5 weeks of Tamoxifen-containing chow (HARLAN LABORATORIES, Indianapolis, Ind., USA) administration followed by 1 week of standard chow. Pulmonary hypertension was induced exposing mice to 3 weeks of hypoxia (10% FiO 2 ). Then, mice were submitted to anesthesia induced by inhalation of isoflurane 3% mixed with air and maintained decreasing isoflurance concentration between 1% and 1.5%. Right-heart catheterization and organ processing were performed using standard methods.
  • Tamoxifen-containing chow HARLAN LABORATORIES, Indianapolis, Ind., USA
  • the heart was taken out the 30 thoracic cage, auricles were removed and right ventricles were separated from left ventricles associated to septa. The weight of each part was measured and the ratio of the right ventricle weigh to the left ventricle with septum weigh was calculated for each mouse. Lungs were processed inflating them with 10 mL of a mixture of saline and OCT 1/1 ratio (ShandonTM CryomatrixTM, THERMOFISCHER SCIENTIFIC). Ventricles and inflated lungs were then frozen in cooled isopentane (VWR) and stored at ⁇ 80° C.
  • VWR isopentane
  • NMDAR activation has been previously associated to aortic smooth muscle cell proliferation depending of MAPK and PI3K signaling pathway activation 25 . Besides, NMDAR activation has been pointed out as an important component of the aberrant proliferation of cancer cells and PAH PASMCs exhibit a cancer-like phenotype.
  • KO mice knockout mice for NMDAR, with targeted deletion of the Grin1 gene in SMCs using a Cre/Lox approach.
  • Grin1 gene expression was clearly reduced in pulmonary arteries from KO mice compared to wild-type (WT) mice indicating efficient genetic recombination.
  • Normoxic KO and WT mice were exposed to 3 weeks normoxia or hypoxia (FiO 2 10%) in order to induce experimental PH.
  • Normoxic KO and WT mice didn't show any differences in right ventricular systolic pressure (RVSP) and right cardiac hypertrophy (Fulton index) opposite to hypoxic KO mice presenting significantly lower RVSP and Fulton index than hypoxic WT mice. This was associated to a decreased muscularization of small pulmonary arterioles ( ⁇ 50 ⁇ m external diameter) in hypoxic KO mice compared to WT mice.
  • RVSP right ventricular systolic pressure
  • Fulton index right cardiac hypertrophy
  • NMDAR uncompetitive antagonists MK-801 and memantine (MMT)
  • MK-801 and MMT memantine
  • PDGF-BB a growth factor of PASMCs
  • PAH vascular remodeling
  • NMDAR antagonists MK-801 and MMT attenuated hPASMCs proliferation induced by PDGF-BB without adding any NMDAR agonists to the medium.
  • PDGF-BB did not further increase the basal release of glutamate from control hPASMCs suggesting a role in mobilizing NMDARs.
  • Kinetics analysis showed increased GluN1 phosphorylation after 10 min of PDGF-BB exposure and followed by a slight decrease after 30 min to 1 h.
  • PDGF-BB could activate NMDAR trafficking, phosphorylating the obligatory GluN1 subunit within minutes, then contributing to proliferative effects.
  • PDGF-BB and/or ET-1 could be responsible for the increased phosphorylation of GluN1 observed in situ in pulmonary arteries from PAH patients especially in PASMC, a cell type known to express abundantly the PDGFR.
  • NMDAR expressed by PASMC contributes to vascular remodeling, resulting in an increased RVSP and subsequently right cardiac hypertrophy in the hypoxic mice model.
  • PDGF-BB could mobilize NMDAR in order to participate to the resulting proliferative effect.
  • NMDAR activation in CNS endothelial cell can disturb the endothelial barrier through production of reactive oxygen species, favor monocyte transmigration and induce proliferation 18-20,22 .
  • vascular remodeling associated to PAH involves breakdown of endothelial junctions, oxidant stress, proliferation and adventitial inflammation 40 .
  • NMDARs potentiated GluN1 protein
  • Synaptic-like contact was defined as a close contact ⁇ 70 ⁇ m between two endothelial cells without junctions.
  • FIG. 2 a the total number of immunogold particles per 100 ⁇ m 2 of endothelial cells
  • FIG. 1 a the partition/distribution between membrane and cytoplasmic GluN1 as well as the proportion of synaptic-like contact length relative to the total membrane length
  • membrane GluN1 located in these contact areas is increased in plexiform lesion compared to intimal lesion and control artery (29.5% of total membrane GluN1 in plexiform lesion vs 7.7% in intimal lesion and 4.1% in control artery).
  • membrane GluN1 proteins are 3.5 fold enriched inside/along synaptic-like contacts compared to the control artery. In plexiform lesion, this index even reaches 7.1 ( FIG. 1 a ).
  • mice were exposed to normoxia or hypoxia for 3 weeks before measuring RVSP and calculating the Fulton index. No difference was observed in body weight although normoxic KO mice were slightly older than WT mice. In normoxic mice, no difference in RVSP or Fulton index between WT and KO mice was highlighted. After hypoxia, KO mice presented a decreased right cardiac hypertrophy and a decreased RVSP ( FIG. 1 c,d ). Morphometric analysis of the pulmonary arteries revealed a significant decrease of the muscularization only in small arterioles ( ⁇ 50 ⁇ m external diameter) between WT and KO mice after chronic hypoxia ( FIG. 1 e ). No difference was observed for larger arteries between WT and KO mice in both normoxia and hypoxia.
  • NMDAR antagonists MK-801 and MMT dose-dependently inhibit hPMVEC proliferation stimulated either by FBS 10% or VEGF-A 10 ng ⁇ ml ⁇ 1 without adding exogenous NMDAR agonists ( FIG. 1 f , FIG. 2 e ).
  • VEGF is one of the main angiogenesis mediators, we have explored the hypothesis whether NMDAR activation is involved in angiogenesis.
  • MK-801 and the competitive NMDAR antagonist DAP-5 decrease total tube length formation and the number of nodes ( FIG. 1 g FIG. 2 f ).
  • MK-801 dose-dependently inhibits the formation of the endothelial tube network ( FIG. 1 h , FIG. 2 g ).
  • NMDAR is dysregulated in endothelial cells in PAH intimal and plexiform lesions and contributes to vascular remodeling. Moreover it participates to VEGF proliferation and angiogenesis.
  • NMDAR-associated synaptic proteins neurologin and neurexin that trigger synapse formation in the CNS have already been described in vascular cells playing a role in angiogenesis 56 and common variants in cerebellin 2, a partner of neurexin and expressed by pulmonary endothelial cells, increases the risk of PAH by approximately two-fold 57 .
  • Stepulak, A. et al. NMDA antagonist inhibits the extracellular signal-regulated kinase pathway and suppresses cancer growth. Proc. Natl. Acad. Sci. U.S.A. 102, 15605-15610 (2005).
  • VEGF modulates NMDA receptors activity in cerebellar granule cells through Src-family kinases before synapse formation. Proc. Natl. Acad. Sci. U.S.A. 108, 13782-13787 (2011).
US15/779,369 2015-11-30 2016-11-30 NMDAR antagonists for the treatment of diseases associated with angiogenesis Active US10583171B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP15306894.5 2015-11-30
EP15306894 2015-11-30
EP15306894 2015-11-30
PCT/EP2016/079340 WO2017093354A1 (en) 2015-11-30 2016-11-30 Nmdar antagonists for the treatment of diseases associated with angiogenesis

Publications (2)

Publication Number Publication Date
US20180318389A1 US20180318389A1 (en) 2018-11-08
US10583171B2 true US10583171B2 (en) 2020-03-10

Family

ID=54782643

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/779,369 Active US10583171B2 (en) 2015-11-30 2016-11-30 NMDAR antagonists for the treatment of diseases associated with angiogenesis

Country Status (4)

Country Link
US (1) US10583171B2 (es)
EP (1) EP3383429B1 (es)
ES (1) ES2843724T3 (es)
WO (1) WO2017093354A1 (es)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024040232A2 (en) * 2022-08-19 2024-02-22 Lankenau Institute For Medical Research Methods of treating conditions using anti-nmda receptor antibodies

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251948B1 (en) 1990-03-02 2001-06-26 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University And The University Of Oregon Tri-and tetra-substituted guanidines and their use as excitatory amino acid antagonists
GB2428675A (en) 2005-07-29 2007-02-07 Arakis Ltd Ifenprodil derivatives
US20080145849A1 (en) 2005-04-19 2008-06-19 North William G Methods for diagnosing and treating neuroendocrine cancer
US20080193503A1 (en) 2004-04-30 2008-08-14 Allergan, Inc. Memantine intravitreal implants
US20100130528A1 (en) 2008-11-21 2010-05-27 Auspex Pharmaceuticals, Inc. Adamantane modulators of nmda receptor and/or 5ht3 receptor
US20100196354A1 (en) * 2007-01-16 2010-08-05 The Johns Hopkins University Glutamate receptor antagonists and methods of use
US20100216775A1 (en) 2007-02-21 2010-08-26 SSV Therapeutics Adamantanamines and Neramexane Salts of Thiomolybdic and Thiotungstic Acids
WO2010121973A1 (en) 2009-04-23 2010-10-28 Universität Zürich Blockers of nmda receptor for the treatment of sickle cell anemia
EP2264035A1 (en) 2009-06-04 2010-12-22 Merz Pharma GmbH & Co. KGaA Glycine B antagonists
US20140212421A1 (en) * 2008-07-10 2014-07-31 Esbatech, An Alcon Biomedical Research Unit Llc Methods and compositions for enhanced delivery of macromolecules
WO2014187879A2 (en) 2013-05-21 2014-11-27 Paion Deutschland Gmbh Novel antibody useful in neurological or neurodegenerative disorders
US20180000802A1 (en) * 2015-06-26 2018-01-04 Institute For Basic Science Pharmaceutical composition for prevention and treatment of mental disease with enhanced nmdar function

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE565262A (es) 1957-09-19
US3097136A (en) 1960-04-04 1963-07-09 Parke Davis & Co Process for producing a depressant-like effect on the central nervous system
US3310469A (en) 1961-08-28 1967-03-21 Du Pont Pharmaceutical compositions and methods utilizing 1-aminoadamantane and its derivatives
US3254124A (en) 1962-06-29 1966-05-31 Parke Davis & Co Aminoketones and methods for their production
US3352912A (en) 1963-07-24 1967-11-14 Du Pont Adamantanes and tricyclo[4. 3. 1. 1 3.8] undecanes
FR5733M (es) 1966-09-27 1968-01-22
US4399141A (en) 1977-09-19 1983-08-16 Merck & Co., Inc. 5-Alkyl or hydroxyalkyl substituted-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imines and anticonvulsant use thereof
NL191488C (nl) 1977-09-19 1995-08-04 Merck & Co Inc Op de 5-plaats gesubstitueerde 10,11-dihydro-5H-dibenzo£a,d|cyclohepteen-5,10-iminen en farmaceutische preparaten die deze bevatten.
IL67646A0 (en) 1982-01-15 1983-05-15 Lilly Co Eli Ascorbic acid ethers and related compounds
FR2534580A1 (fr) 1982-10-13 1984-04-20 Synthelabo Derives de phenyl-1 piperidino-2 propanol, leur preparation, et medicaments qui les contiennent
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4551552A (en) 1984-05-23 1985-11-05 E. I. Du Pont De Nemours And Company Process for preparing rimantadine
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
DE10299048I2 (de) 1989-04-14 2006-07-13 Merz Pharma Gmbh & Co Kgaa Verwendung von Adamantan-Derivaten zur Pr{vention und Behandlung der cerebralen Isch{mie
JPH05507717A (ja) 1990-06-25 1993-11-04 ザ・デュポン・メルク・ファーマシュウティカル・カンパニー 軟ゼラチンカプセル剤中で使用するための高められた溶解特性を有するアマンタジン塩酸塩懸濁液
EP0859773A1 (en) 1995-09-15 1998-08-26 Pfizer Inc. Phenol derivatives with pharmaceutical activity
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
AUPP249298A0 (en) 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US7595297B2 (en) 1999-06-02 2009-09-29 Michael Tymianski Method of reducing injury to mammalian cells
US7510824B2 (en) 1999-06-02 2009-03-31 Nono Inc. Method of screening peptides useful in treating traumatic injury to the brain or spinal cord
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
US6916816B2 (en) 1999-12-16 2005-07-12 The Board Of Regents Of The University Of Nebraska Phenanthryl piperazinyl dicarboxylic acids as selective NMDA receptor modulating agents
WO2001068836A2 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
JP2005506292A (ja) 2001-03-08 2005-03-03 エモリー ユニバーシティ pHに依存するNMDAレセプターアンタゴニスト
US7786140B2 (en) 2003-09-25 2010-08-31 Shionogi & Co., Ltd. Piperidine derivative having NMDA receptor antagonistic activity
US20060063802A1 (en) 2004-03-29 2006-03-23 Matthieu Guitton Methods for the treatment of tinnitus induced by cochlear excitotoxicity
WO2006020171A1 (en) 2004-07-19 2006-02-23 University Of Florida Research Foundation, Inc. Methods and materials for treating mental illness
CN1993363A (zh) 2004-08-03 2007-07-04 默克公司 1,3-二取代的杂芳基nmda/nr2b拮抗剂
EP1791569A4 (en) 2004-08-23 2009-12-16 Univ Emory IMPROVED SELECTION OF PH-DEPENDENT COMPOUNDS FOR IN VIVO THERAPY
WO2006113471A2 (en) 2005-04-19 2006-10-26 Merck & Co., Inc. N-alkyl-azacycloalkyl nmda/nr2b antagonists
WO2007126020A1 (ja) 2006-04-28 2007-11-08 National University Corporation Chiba University Nmda受容体チャネルブロック作用を示す化合物及びそれを用いた薬剤
WO2007149508A2 (en) 2006-06-19 2007-12-27 The University Of The Sciences In Philadelphia Non-competitive nmda receptor antagonists
WO2008134525A1 (en) 2007-04-26 2008-11-06 Auspex Pharmaeuticals, Inc. Deuterium labelled ketamine
CA2809739C (en) 2007-05-01 2016-11-15 Concert Pharmaceuticals Inc. Morphinan compounds
CA2693032A1 (en) 2007-06-29 2009-01-08 Emory University Nmda receptor antagonists for neuroprotection
US8008253B2 (en) 2007-07-03 2011-08-30 Andrew Tasker Treatment for anxiety
EA201070571A1 (ru) 2007-11-06 2010-12-30 Эмори Юниверсити Улучшенные способы идентификации антагонистов nmda-рецептора
GB2470495B (en) 2008-01-16 2012-12-26 Biotechnology Res Corp Ltd Oxazolidine derivatives as NMDA antagonists
WO2009129181A1 (en) 2008-04-14 2009-10-22 Concert Pharmaceuticals Inc. Propanediol-dicarbamate derivatives
BRPI0912362A2 (pt) 2008-05-09 2015-10-06 Univ Emory antagonista do receptor nmda para o tratamento de transtornos neuropsiquiátricos
ES2446306T3 (es) 2008-05-16 2014-03-07 Nono Inc. Uso de un inhibidor de psd-95 tratamiento para la epilepsia
CA2727141A1 (en) 2008-06-13 2009-12-17 Centre For Addiction And Mental Health Compositions and methods for modulating nicotinic/nmda receptor function
CN101337555B (zh) 2008-07-29 2010-07-28 好孩子儿童用品有限公司 可折叠的童车及其折叠方法
CN102076738B (zh) 2008-07-30 2012-08-29 Sk新技术株式会社 新型配位络合物和使用该络合物作为催化剂通过二氧化碳和环氧化物的共聚合制备聚碳酸酯的方法
JP5702288B2 (ja) 2008-09-18 2015-04-15 ノースウエスタン ユニバーシティ Nmdaレセプターモジュレータ及びその用途
EP2334678B1 (en) 2008-09-19 2012-12-26 Concert Pharmaceuticals Inc. Morphinan compounds
TW201026667A (en) 2008-09-30 2010-07-16 Merz Pharma Gmbh & Co Kgaa Glycine b antagonists
TWI380992B (zh) 2010-03-11 2013-01-01 Univ Ishou 一種抑制nmda受體nr1之小段干擾rna、該小段干擾rna用以抑制皮下組織nmda受體nr1之方法、該小段干擾rna之用途以及一種減緩皮膚炎症性疼痛之藥物化合物
WO2012019106A2 (en) 2010-08-06 2012-02-09 Board Of Regents Of The University Of Nebraska Positive and negative modulators of nmda receptors

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251948B1 (en) 1990-03-02 2001-06-26 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University And The University Of Oregon Tri-and tetra-substituted guanidines and their use as excitatory amino acid antagonists
US20080193503A1 (en) 2004-04-30 2008-08-14 Allergan, Inc. Memantine intravitreal implants
US20080145849A1 (en) 2005-04-19 2008-06-19 North William G Methods for diagnosing and treating neuroendocrine cancer
GB2428675A (en) 2005-07-29 2007-02-07 Arakis Ltd Ifenprodil derivatives
US20100196354A1 (en) * 2007-01-16 2010-08-05 The Johns Hopkins University Glutamate receptor antagonists and methods of use
US20100216775A1 (en) 2007-02-21 2010-08-26 SSV Therapeutics Adamantanamines and Neramexane Salts of Thiomolybdic and Thiotungstic Acids
US20140212421A1 (en) * 2008-07-10 2014-07-31 Esbatech, An Alcon Biomedical Research Unit Llc Methods and compositions for enhanced delivery of macromolecules
US20100130528A1 (en) 2008-11-21 2010-05-27 Auspex Pharmaceuticals, Inc. Adamantane modulators of nmda receptor and/or 5ht3 receptor
WO2010121973A1 (en) 2009-04-23 2010-10-28 Universität Zürich Blockers of nmda receptor for the treatment of sickle cell anemia
EP2264035A1 (en) 2009-06-04 2010-12-22 Merz Pharma GmbH & Co. KGaA Glycine B antagonists
WO2014187879A2 (en) 2013-05-21 2014-11-27 Paion Deutschland Gmbh Novel antibody useful in neurological or neurodegenerative disorders
US20180000802A1 (en) * 2015-06-26 2018-01-04 Institute For Basic Science Pharmaceutical composition for prevention and treatment of mental disease with enhanced nmdar function

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Fares et al.; "Pulmonary hypertension: diagnostic and therapeutic challenges"; Therapeutics and Clinical risk Vlanagement, vol. 11, Aug. 1, 2015, pp. 1221-1233.
Williams et al.; "Ketamine Does Not Increase Pulmonary Vascular Resistance in Children with Pulmonary Hypertension Undergoing Sevoflurane Anesthesia and Spontaneous Ventilation"; Anesthesia and Analgesia, vol. 105, No. 6, Dec. 1, 2007, pp. 1578-1584.

Also Published As

Publication number Publication date
WO2017093354A1 (en) 2017-06-08
EP3383429A1 (en) 2018-10-10
US20180318389A1 (en) 2018-11-08
ES2843724T3 (es) 2021-07-20
EP3383429B1 (en) 2020-10-14

Similar Documents

Publication Publication Date Title
US9937224B2 (en) Modulation of axon degeneration
JP5859307B2 (ja) 眼の血管新生を阻害する方法
US20160199457A1 (en) Modulation of axon degeneration
Prakash et al. Inhibition of renal rho kinase attenuates ischemia/reperfusion-induced injury
Sasaki et al. ATP-binding cassette transporter A subfamily 8 is a sinusoidal efflux transporter for cholesterol and taurocholate in mouse and human liver
JP6689484B2 (ja) 網膜炎症の治療において使用するための薬剤
US10583171B2 (en) NMDAR antagonists for the treatment of diseases associated with angiogenesis
US20180298104A1 (en) Methods and pharmaceutical compositions for the treatment of th17 mediated diseases
WO2011076946A2 (en) Methods and compositions for the treatment of alzheimer
Triana-Baltzer et al. Pre-and postsynaptic actions of L1-CAM in nicotinic pathways
US20080206253A1 (en) Method For Preventing or Treating Pain in a Mammal
US20230312751A1 (en) Anti-Fibrotic Combination
WO2024028476A1 (en) Methods for the treatment of th2-mediated diseases
WO2020016160A1 (en) Method to treat neurological diseases
US20120328631A1 (en) Ep1 inhibition
CN106102766A (zh) 用于治疗视网膜炎症的试剂
JP2017524739A (ja) 神経筋接合部関連疾患の処置方法

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: INSERM (INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE), FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COHEN-KAMINSKY, SYLVIA;DUMAS, SEBASTIEN;BRU-MERCIER, GILLES;SIGNING DATES FROM 20180820 TO 20180827;REEL/FRAME:046789/0703

Owner name: INSERM (INSTITUT NATIONAL DE LA SANTE ET DE LA REC

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COHEN-KAMINSKY, SYLVIA;DUMAS, SEBASTIEN;BRU-MERCIER, GILLES;SIGNING DATES FROM 20180820 TO 20180827;REEL/FRAME:046789/0703

Owner name: UNIVERSITE PARIS-SUD, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COHEN-KAMINSKY, SYLVIA;DUMAS, SEBASTIEN;BRU-MERCIER, GILLES;SIGNING DATES FROM 20180820 TO 20180827;REEL/FRAME:046789/0703

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP., ISSUE FEE NOT PAID

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP, ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4